1
|
Molnar T, Lehoczki A, Fekete M, Varnai R, Zavori L, Erdo-Bonyar S, Simon D, Berki T, Csecsei P, Ezer E. Mitochondrial dysfunction in long COVID: mechanisms, consequences, and potential therapeutic approaches. GeroScience 2024; 46:5267-5286. [PMID: 38668888 PMCID: PMC11336094 DOI: 10.1007/s11357-024-01165-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/15/2024] [Indexed: 08/22/2024] Open
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has introduced the medical community to the phenomenon of long COVID, a condition characterized by persistent symptoms following the resolution of the acute phase of infection. Among the myriad of symptoms reported by long COVID sufferers, chronic fatigue, cognitive disturbances, and exercise intolerance are predominant, suggesting systemic alterations beyond the initial viral pathology. Emerging evidence has pointed to mitochondrial dysfunction as a potential underpinning mechanism contributing to the persistence and diversity of long COVID symptoms. This review aims to synthesize current findings related to mitochondrial dysfunction in long COVID, exploring its implications for cellular energy deficits, oxidative stress, immune dysregulation, metabolic disturbances, and endothelial dysfunction. Through a comprehensive analysis of the literature, we highlight the significance of mitochondrial health in the pathophysiology of long COVID, drawing parallels with similar clinical syndromes linked to post-infectious states in other diseases where mitochondrial impairment has been implicated. We discuss potential therapeutic strategies targeting mitochondrial function, including pharmacological interventions, lifestyle modifications, exercise, and dietary approaches, and emphasize the need for further research and collaborative efforts to advance our understanding and management of long COVID. This review underscores the critical role of mitochondrial dysfunction in long COVID and calls for a multidisciplinary approach to address the gaps in our knowledge and treatment options for those affected by this condition.
Collapse
Affiliation(s)
- Tihamer Molnar
- Department of Anaesthesiology and Intensive Care, Medical School, University of Pecs, Pecs, Hungary
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Haematology and Stem Cell Transplantation, National Institute for Haematology and Infectious Diseases, South Pest Central Hospital, 1097, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Reka Varnai
- Department of Primary Health Care, Medical School University of Pecs, Pecs, Hungary
| | | | - Szabina Erdo-Bonyar
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Pecs, Hungary
| | - Diana Simon
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Pecs, Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Pecs, Hungary
| | - Peter Csecsei
- Department of Neurosurgery, Medical School, University of Pecs, Ret U 2, 7624, Pecs, Hungary.
| | - Erzsebet Ezer
- Department of Anaesthesiology and Intensive Care, Medical School, University of Pecs, Pecs, Hungary
| |
Collapse
|
2
|
Correa Segura F, Macías Macías FI, Velázquez Delgado KA, Ramos-Godinez MDP, Ruiz-Ramírez A, Flores P, Huerta-García E, López-Marure R. Food-grade titanium dioxide (E171) and zinc oxide nanoparticles induce mitochondrial permeability and cardiac damage after oral exposure in rats. Nanotoxicology 2024; 18:122-133. [PMID: 38436290 DOI: 10.1080/17435390.2024.2323069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
Food-grade titanium dioxide (E171) and zinc oxide nanoparticles (ZnO NPs) are found in diverse products for human use. E171 is used as whitening agent in food and cosmetics, and ZnO NPs in food packaging. Their potential multi-organ toxicity has raised concerns on their safety. Since mitochondrial dysfunction is a key aspect of cardio-pathologies, here, we evaluate the effect of chronic exposure to E171 and ZnO NPs in rats on cardiac mitochondria. Changes in cardiac electrophysiology and body weight were measured. E171 reduced body weight more than 10% after 5 weeks. Both E171 and ZnO NPs increased systolic blood pressure (SBP) from 110-120 to 120-140 mmHg after 45 days of treatment. Both NPs altered the mitochondrial permeability transition pore (mPTP), reducing calcium requirement for permeability by 60% and 93% in E171- and ZnO NPs-exposed rats, respectively. Treatments also affected conformational state of adenine nucleotide translocase (ANT). E171 reduced the binding of EMA to Cys 159 in 30% and ZnO NPs in 57%. Mitochondrial aconitase activity was reduced by roughly 50% with both NPs, indicating oxidative stress. Transmission electron microscopy (TEM) revealed changes in mitochondrial morphology including sarcomere discontinuity, edema, and hypertrophy in rats exposed to both NPs. In conclusion, chronic oral exposure to NPs induces functional and morphological damage in cardiac mitochondria, with ZnO NPs being more toxic than E171, possibly due to their dissociation in free Zn2+ ion form. Therefore, chronic intake of these food additives could increase risk of cardiovascular disease.
Collapse
Affiliation(s)
- Francisco Correa Segura
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | | | | | | | - Angélica Ruiz-Ramírez
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Pedro Flores
- Departamento de Instrumentación, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Elizabeth Huerta-García
- División Académica Multidisciplinaria de Jalpa de Méndez, Universidad Juárez Autónoma de Tabasco, Villahermosa, México
| | - Rebeca López-Marure
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| |
Collapse
|
3
|
Piao L, Fang YH, Fisher M, Hamanaka RB, Ousta A, Wu R, Mutlu GM, Garcia AJ, Archer SL, Sharp WW. Dynamin-related protein 1 is a critical regulator of mitochondrial calcium homeostasis during myocardial ischemia/reperfusion injury. FASEB J 2024; 38:e23379. [PMID: 38133921 DOI: 10.1096/fj.202301040rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Dynamin-related protein 1 (Drp1) is a cytosolic GTPase protein that when activated translocates to the mitochondria, meditating mitochondrial fission and increasing reactive oxygen species (ROS) in cardiomyocytes. Drp1 has shown promise as a therapeutic target for reducing cardiac ischemia/reperfusion (IR) injury; however, the lack of specificity of some small molecule Drp1 inhibitors and the reliance on the use of Drp1 haploinsufficient hearts from older mice have left the role of Drp1 in IR in question. Here, we address these concerns using two approaches, using: (a) short-term (3 weeks), conditional, cardiomyocyte-specific, Drp1 knockout (KO) and (b) a novel, highly specific Drp1 GTPase inhibitor, Drpitor1a. Short-term Drp1 KO mice exhibited preserved exercise capacity and cardiac contractility, and their isolated cardiac mitochondria demonstrated increased mitochondrial complex 1 activity, respiratory coupling, and calcium retention capacity compared to controls. When exposed to IR injury in a Langendorff perfusion system, Drp1 KO hearts had preserved contractility, decreased reactive oxygen species (ROS), enhanced mitochondrial calcium capacity, and increased resistance to mitochondrial permeability transition pore (MPTP) opening. Pharmacological inhibition of Drp1 with Drpitor1a following ischemia, but before reperfusion, was as protective as Drp1 KO for cardiac function and mitochondrial calcium homeostasis. In contrast to the benefits of short-term Drp1 inhibition, prolonged Drp1 ablation (6 weeks) resulted in cardiomyopathy. Drp1 KO hearts were also associated with decreased ryanodine receptor 2 (RyR2) protein expression and pharmacological inhibition of the RyR2 receptor decreased ROS in post-IR hearts suggesting that changes in RyR2 may have a role in Drp1 KO mediated cardioprotection. We conclude that Drp1-mediated increases in myocardial ROS production and impairment of mitochondrial calcium handling are key mechanisms of IR injury. Short-term inhibition of Drp1 is a promising strategy to limit early myocardial IR injury which is relevant for the therapy of acute myocardial infarction, cardiac arrest, and heart transplantation.
Collapse
Affiliation(s)
- Lin Piao
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yong-Hu Fang
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Michael Fisher
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Robert B Hamanaka
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Alaa Ousta
- Department of Emergency Medicine, Duke University, Durham, North Carolina, USA
| | - Rongxu Wu
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Institute for Integrative Physiology, University of Chicago, Chicago, Illinois, USA
| | - Alfredo J Garcia
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Institute for Integrative Physiology, University of Chicago, Chicago, Illinois, USA
- The University of Chicago Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Willard W Sharp
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Institute for Integrative Physiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
4
|
Guo L, Wang L, Qin G, Zhang J, Peng J, Li L, Chen X, Wang D, Qiu J, Wang E. M-type pyruvate kinase 2 (PKM2) tetramerization alleviates the progression of right ventricle failure by regulating oxidative stress and mitochondrial dynamics. J Transl Med 2023; 21:888. [PMID: 38062516 PMCID: PMC10702013 DOI: 10.1186/s12967-023-04780-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Right ventricle failure (RVF) is a progressive heart disease that has yet to be fully understood at the molecular level. Elevated M-type pyruvate kinase 2 (PKM2) tetramerization alleviates heart failure, but detailed molecular mechanisms remain unclear. OBJECTIVE We observed changes in PKM2 tetramerization levels during the progression of right heart failure and in vitro cardiomyocyte hypertrophy and explored the causal relationship between altered PKM2 tetramerization and the imbalance of redox homeostasis in cardiomyocytes, as well as its underlying mechanisms. Ultimately, our goal was to propose rational intervention strategies for the treatment of RVF. METHOD We established RVF in Sprague Dawley (SD) rats by intraperitoneal injection of monocrotaline (MCT). The pulmonary artery pressure and right heart function of rats were assessed using transthoracic echocardiography combined with right heart catheterization. TEPP-46 was used both in vivo and in vitro to promote PKM2 tetramerization. RESULTS We observed that oxidative stress and mitochondrial disorganization were associated with increased apoptosis in the right ventricular tissue of RVF rats. Quantitative proteomics revealed that PKM2 was upregulated during RVF and negatively correlated with the cardiac function. Facilitating PKM2 tetramerization promoted mitochondrial network formation and alleviated oxidative stress and apoptosis during cardiomyocyte hypertrophy. Moreover, enhancing PKM2 tetramer formation improved cardiac mitochondrial morphology, mitigated oxidative stress and alleviated heart failure. CONCLUSION Disruption of PKM2 tetramerization contributed to RVF by inducing mitochondrial fragmentation, accumulating ROS, and finally promoted the progression of cardiomyocyte apoptosis. Facilitating PKM2 tetramerization holds potential as a promising therapeutic approach for RVF.
Collapse
Affiliation(s)
- Lizhe Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Gang Qin
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Junjie Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jin Peng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Longyan Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Dandan Wang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
| | - Jian Qiu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China.
- Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China.
| |
Collapse
|
5
|
Yang C, Cheng J, Zhu Q, Pan Q, Ji K, Li J. Review of the Protective Mechanism of Paeonol on Cardiovascular Disease. Drug Des Devel Ther 2023; 17:2193-2208. [PMID: 37525853 PMCID: PMC10387245 DOI: 10.2147/dddt.s414752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 08/02/2023] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death in the world. Paeonol(Pae) is a phenolic component extracted from peony bark, peony root and Xu Changqing. Studies have shown that Pae can protect cardiomyocytes by inhibiting oxidative stress, promoting mitochondrial fusion, regulating mitochondrial autophagy and inhibiting inflammation. In addition, Pae improves ventricular remodeling by inhibiting myocardial apoptosis, hypertrophy and fibrosis. Pae also has a good protective effect on blood vessels by inhibiting vascular inflammation, reducing the expression of adhesion molecules, inhibiting vascular proliferation, and inhibiting oxidative stress and endoplasmic reticulum stress(ERS). Pae also has the effect of anti-endothelial cell senescence, promoting thrombus recanalization and vasodilating. In conclusion, the molecular targets of Pae are very complex, and the relationship between different targets and signaling pathways cannot be clearly explained, which requires us to use systems biology methods to further study specific molecular targets of Pae. It has to be mentioned that the bioavailability of Pae is poor, and some nanotechnology-assisted drug delivery systems improve the therapeutic effect of Pae. We reviewed the protective mechanism of paeonol on the cardiovascular system, hoping to provide help for drug development in the treatment of CVD.
Collapse
Affiliation(s)
- Chunkun Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Jiawen Cheng
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, People's Republic of China
| | - Jun Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
6
|
4-Hydroxycinnamic acid attenuates neuronal cell death by inducing expression of plasma membrane redox enzymes and improving mitochondrial functions. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
7
|
Salimi A, Shabani M, Bayrami D, Saray A, Farshbaf Moghimi N. Gallic acid and sesame oil exert cardioprotection via mitochondrial protection and antioxidant properties on Ketamine-Induced cardiotoxicity model in rats. TOXIN REV 2023. [DOI: 10.1080/15569543.2023.2165503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences
| | - Mohammad Shabani
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Deniz Bayrami
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Armin Saray
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nastaran Farshbaf Moghimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
8
|
Voglhuber J, Holzer M, Radulović S, Thai PN, Djalinac N, Matzer I, Wallner M, Bugger H, Zirlik A, Leitinger G, Dedkova EN, Bers DM, Ljubojevic-Holzer S. Functional remodelling of perinuclear mitochondria alters nucleoplasmic Ca 2+ signalling in heart failure. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210320. [PMID: 36189813 PMCID: PMC9527904 DOI: 10.1098/rstb.2021.0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/23/2022] [Indexed: 11/12/2022] Open
Abstract
Mitochondrial dysfunction in cardiomyocytes is a hallmark of heart failure development. Although initial studies recognized the importance of different mitochondrial subpopulations, there is a striking lack of direct comparison of intrafibrillar (IF) versus perinuclear (PN) mitochondria during the development of HF. Here, we use multiple approaches to examine the morphology and functional properties of IF versus PN mitochondria in pressure overload-induced cardiac remodelling in mice, and in non-failing and failing human cardiomyocytes. We demonstrate that PN mitochondria from failing cardiomyocytes are more susceptible to depolarization of mitochondrial membrane potential, reactive oxygen species generation and impairment in Ca2+ uptake compared with IF mitochondria at baseline and under physiological stress protocol. We also demonstrate, for the first time to our knowledge, that under normal conditions PN mitochondrial Ca2+ uptake shapes nucleoplasmic Ca2+ transients (CaTs) and limits nucleoplasmic Ca2+ loading. The loss of PN mitochondrial Ca2+ buffering capacity translates into increased nucleoplasmic CaTs and may explain disproportionate rise in nucleoplasmic [Ca2+] in failing cardiomyocytes at increased stimulation frequencies. Therefore, a previously unidentified benefit of restoring the mitochondrial Ca2+ uptake may be normalization of nuclear Ca2+ signalling and alleviation of altered excitation-transcription, which could be an important therapeutic approach to prevent adverse cardiac remodelling. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Julia Voglhuber
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Michael Holzer
- BioTechMed-Graz, Graz, Austria
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Snježana Radulović
- Research Unit Electron Microscopic Techniques, Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Phung N. Thai
- Department of Internal Medicine, Cardiovascular Medicine, University of California Davis, Davis, CA, USA
| | - Natasa Djalinac
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Ingrid Matzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Markus Wallner
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, PA, USA
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Research Unit Electron Microscopic Techniques, Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Elena N. Dedkova
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Department of Molecular Biosciences, University of California Davis, Davis, CA, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
9
|
Paeonol protects against doxorubicin-induced cardiotoxicity by promoting Mfn2-mediated mitochondrial fusion through activating the PKCε-Stat3 pathway. J Adv Res 2022; 47:151-162. [PMID: 35842187 PMCID: PMC10173194 DOI: 10.1016/j.jare.2022.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 07/10/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION The anti-cancer medication doxorubicin (Dox) is largely restricted in clinical usage due to its significant cardiotoxicity. The only medication approved by the FDA for Dox-induced cardiotoxicity is dexrazoxane, while it may reduce the sensitivity of cancer cells to chemotherapy and is restricted for use. There is an urgent need for the development of safe and effective medicines to alleviate Dox-induced cardiotoxicity. OBJECTIVES The objective of this study was to determine whether Paeonol (Pae) has the ability to protect against Dox-induced cardiotoxicity and if so, what are the underlying mechanisms involved. METHODS Sprague-Dawley rats and primary cardiomyocytes were used to create Dox-induced cardiotoxicity models. Pae's effects on myocardial damage, mitochondrial function, mitochondrial dynamics and signaling pathways were studied using a range of experimental methods. RESULTS Pae enhanced Mfn2-mediated mitochondrial fusion, restored mitochondrial function and cardiac performance both in vivo and in vitro under the Dox conditions. The protective properties of Pae were blunted when Mfn2 was knocked down or knocked out in Dox-induced cardiomyocytes and hearts respectively. Mechanistically, Pae promoted Mfn2-mediated mitochondria fusion by activating the transcription factor Stat3, which bound to the Mfn2 promoter in a direct manner and up-regulated its transcriptional expression. Furthermore, molecular docking, surface plasmon resonance and co-immunoprecipitation studies showed that Pae's direct target was PKCε, which interacted with Stat3 and enabled its phosphorylation and activation. Pae-induced Stat3 phosphorylation and Mfn2-mediated mitochondrial fusion were inhibited when PKCε was knocked down. Furthermore, Pae did not interfere with Dox's antitumor efficacy in several tumor cells. CONCLUSION Pae protects the heart against Dox-induced damage by stimulating mitochondrial fusion via the PKCε-Stat3-Mfn2 pathway, indicating that Pae might be a promising therapeutic therapy for Dox-induced cardiotoxicity while maintaining Dox's anticancer activity.
Collapse
|
10
|
Abstract
Although currently employed therapies for heart failure decrease overall mortality and improve patient quality of life temporarily, the disease is known to progress even for patients who receive all guideline-recommended therapies. This indicates that our concise understanding of heart failure and of disease progression is incomplete, and there is a need for new interventions that may augment, or even supplant, currently available options. A literature review reveals that an exciting, novel area of current research is focused on mitochondria, which are uniquely juxtaposed at the sites of both generation of high-energy molecules and initiation of programmed cell death. Elamipretide is being studied both to maintain cellular biogenetics and prevent reactive oxygen species-induced cell damage by targeting and stabilizing the cardiolipin-cytochrome c supercomplex. Thus far, elamipretide has been shown to increase left ventricular ejection fraction in dog models of heart failure with reduced ejection fraction and to prevent left ventricular remodeling in rats. In early-phase clinical trials, elamipretide administration has not resulted in any severe adverse events, and it has shown promising improvements in cardiac hemodynamics at highest doses. Nonetheless, additional studies are necessary to describe the long-term safety and efficacy of elamipretide.
Collapse
|
11
|
Messerer J, Wrede C, Schipke J, Brandenberger C, Abdellatif M, Eisenberg T, Madeo F, Sedej S, Mühlfeld C. Spermidine supplementation influences mitochondrial number and morphology in the heart of aged mice. J Anat 2021; 242:91-101. [PMID: 34958481 PMCID: PMC9773166 DOI: 10.1111/joa.13618] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/18/2023] Open
Abstract
Aging is associated with cardiac hypertrophy and progressive decline in heart function. One of the hallmarks of cellular aging is the dysfunction of mitochondria. These organelles occupy around 1/4 to 1/3 of the cardiomyocyte volume. During cardiac aging, the removal of defective or dysfunctional mitochondria by mitophagy as well as the dynamic equilibrium between mitochondrial fusion and fission is distorted. Here, we hypothesized that these changes affect the number of mitochondria and alter their three-dimensional (3D) characteristics in aged mouse hearts. The polyamine spermidine stimulates both mitophagy and mitochondrial biogenesis, and these are associated with improved cardiac function and prolonged lifespan. Therefore, we speculated that oral spermidine administration normalizes the number of mitochondria and their 3D morphology in aged myocardium. Young (4-months old) and old (24-months old) mice, treated or not treated with spermidine, were used in this study (n = 10 each). The number of mitochondria in the left ventricles was estimated by design-based stereology using the Euler-Poincaré characteristic based on a disector at the transmission electron microscopic level. The 3D morphology of mitochondria was investigated by 3D reconstruction (using manual contour drawing) from electron microscopic z-stacks obtained by focused ion beam scanning electron microscopy. The volume of the left ventricle and cardiomyocytes were significantly increased in aged mice with or without spermidine treatment. Although the number of mitochondria was similar in young and old control mice, it was significantly increased in aged mice treated with spermidine. The interfibrillar mitochondria from old mice exhibited a lower degree of organization and a greater variation in shape and size compared to young animals. The mitochondrial alignment along the myofibrils in the spermidine-treated mice appeared more regular than in control aged mice, however, old mitochondria from animals fed spermidine also showed a greater diversity of shape and size than young mitochondria. In conclusion, mitochondria of the aged mouse left ventricle exhibited changes in number and 3D ultrastructure that is likely the structural correlate of dysfunctional mitochondrial dynamics. Spermidine treatment reduced, at least in part, these morphological changes, indicating a beneficial effect on cardiac mitochondrial alterations associated with aging.
Collapse
Affiliation(s)
- Jil Messerer
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
| | - Christoph Wrede
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Research Core Unit Electron MicroscopyHannover Medical SchoolHannoverGermany
| | - Julia Schipke
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Biomedical Research in Endstage and Obstructive Lung Disease HannoverMember of the German Center for Lung Research (DZL)HannoverGermany
| | - Christina Brandenberger
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Biomedical Research in Endstage and Obstructive Lung Disease HannoverMember of the German Center for Lung Research (DZL)HannoverGermany
| | | | - Tobias Eisenberg
- BioTechMed GrazGrazAustria,Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria,Field of Excellence BioHealth—University of GrazGrazAustria
| | - Frank Madeo
- BioTechMed GrazGrazAustria,Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria,Field of Excellence BioHealth—University of GrazGrazAustria
| | - Simon Sedej
- Department of CardiologyMedical University of GrazGrazAustria,BioTechMed GrazGrazAustria,Faculty of MedicineUniversity of MariborMariborSlovenia
| | - Christian Mühlfeld
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Research Core Unit Electron MicroscopyHannover Medical SchoolHannoverGermany,Biomedical Research in Endstage and Obstructive Lung Disease HannoverMember of the German Center for Lung Research (DZL)HannoverGermany
| |
Collapse
|
12
|
Liu C, Han Y, Gu X, Li M, Du Y, Feng N, Li J, Zhang S, Maslov LN, Wang G, Pei J, Fu F, Ding M. Paeonol promotes Opa1-mediated mitochondrial fusion via activating the CK2α-Stat3 pathway in diabetic cardiomyopathy. Redox Biol 2021; 46:102098. [PMID: 34418601 PMCID: PMC8385203 DOI: 10.1016/j.redox.2021.102098] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 12/02/2022] Open
Abstract
Diabetes disrupts mitochondrial function and often results in diabetic cardiomyopathy (DCM). Paeonol is a bioactive compound that has been reported to have pharmacological potential for cardiac and mitochondrial protection. This study aims to explore the effects of paeonol on mitochondrial disorderes in DCM and the underlying mechanisms. We showed that paeonol promoted Opa1-mediated mitochondrial fusion, inhibited mitochondrial oxidative stress, and preserved mitochondrial respiratory capacity and cardiac performance in DCM in vivo and in vitro. Knockdown of Opa1 blunted the above protective effects of paeonol in both diabetic hearts and high glucose-treated cardiomyocytes. Mechanistically, inhibitor screening, siRNA knockdown and chromatin immunoprecipitation experiments showed that paeonol-promoted Opa1-mediated mitochondrial fusion required the activation of Stat3, which directly bound to the promoter of Opa1 to upregulate its transcriptional expression. Moreover, pharmmapper screening and molecular docking studies revealed that CK2α served as a direct target of paeonol that interacted with Jak2 and induced the phosphorylation and activation of Jak2-Stat3. Knockdown of CK2α blunted the promoting effect of paeonol on Jak2-Stat3 phosphorylation and Opa1-mediated mitochondrial fusion. Collectively, we have demonstrated for the first time that paeonol is a novel mitochondrial fusion promoter in protecting against hyperglycemia-induced mitochondrial oxidative injury and DCM at least partially via an Opa1-mediated mechanism, a process in which paeonol interacts with CK2α and restores its kinase activity that subsequently increasing Jak2-Stat3 phosphorylation and enhancing the transcriptional level of Opa1. These findings suggest that paeonol or the promotion of mitochondrial fusion might be a promising strategy for the treatment of DCM.
Collapse
Affiliation(s)
- Chaoyang Liu
- Department of Geriatrics Cardiology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yuehu Han
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Man Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yanyan Du
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Na Feng
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Leonid N Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Tomsk, 634000, Russia
| | - Guoen Wang
- Department of Geriatrics Cardiology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Mingge Ding
- Department of Geriatrics Cardiology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
13
|
Fu F, Liu C, Shi R, Li M, Zhang M, Du Y, Wang Q, Li J, Wang G, Pei J, Ding M. Punicalagin Protects Against Diabetic Cardiomyopathy by Promoting Opa1-Mediated Mitochondrial Fusion via Regulating PTP1B-Stat3 Pathway. Antioxid Redox Signal 2021; 35:618-641. [PMID: 33906428 DOI: 10.1089/ars.2020.8248] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aims: This study aims to explore the efficacy of punicalagin (PG) on diabetic cardiomyopathy (DCM), with a specific focus on the mechanisms underlying the effects of PG on mitochondrial fusion/fission dynamics. Results: Cardiac structural and functional abnormalities were ameliorated in diabetic rats receiving PG administration as evidenced by increased ejection fraction, and attenuated myocardial fibrosis and hypertrophy. PG enhanced mitochondrial function and inhibited mitochondria-derived oxidative stress by promoting Opa1-mediated mitochondrial fusion. The benefits of PG could be abrogated by knockdown of Opa1 in vivo and in vitro. Inhibitor screening and chromatin immunoprecipitation analysis showed that Stat3 directly regulated the transcriptional expression of Opa1 by binding to its promoter and was responsible for PG-induced Opa1-mediated mitochondrial fusion. Moreover, pharmmapper screening and molecular docking studies revealed that PG embedded into the activity pocket of PTP1B and inhibited the activity of PTP1B. Overexpression of PTP1B blocked the promoting effect of PG on Stat3 phosphorylation and Opa1-mediated mitochondrial fusion, whereas knockdown of PTP1B mimicked the benefits of PG in high-glucose-treated cardiomyocytes. Innovation: Our study is the first to identify PG as a novel mitochondrial fusion promoter against hyperglycemia-induced mitochondrial oxidative injury and cardiomyopathy by upregulating Opa1 via regulating PTP1B-Stat3 pathway. Conclusion: PG protects against DCM by promoting Opa1-mediated mitochondrial fusion, a process in which PG interacts with PTP1B and inhibits its activity, which in turn increases Stat3 phosphorylation and then enhances the transcriptional expression of Opa1. These results suggest that PG might be a promising new therapeutic approach against diabetic cardiac complication. Antioxid. Redox Signal. 35, 618-641.
Collapse
Affiliation(s)
- Feng Fu
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Chaoyang Liu
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Rui Shi
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Man Li
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Min Zhang
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yanyan Du
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Qiaojuan Wang
- School of Life Sciences, Northwest University, Xi'an, China.,Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jun Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Guoen Wang
- Department of Geriatrics Cardiology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Mingge Ding
- Department of Geriatrics Cardiology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Luo T, Liu H, Chen B, Liu H, Abdel-Latif A, Kitakaze M, Wang X, Wu Y, Chou D, Kim JK. A novel role of claudin-5 in prevention of mitochondrial fission against ischemic/hypoxic stress in cardiomyocytes. Can J Cardiol 2021; 37:1593-1606. [PMID: 33838228 DOI: 10.1016/j.cjca.2021.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/20/2021] [Accepted: 03/29/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Downregulation of claudin-5 in the heart is associated with the end-stage heart failure. However, the underlying mechanism of claudin-5 is unclear. Here we investigated the molecular actions of claudin-5 in perspective of mitochondria in cardiomyocytes to better understand the role of claudin-5 in cardioprotection during ischemia. METHODS AND RESULTS Claudin-5 was detected in the murine heart tissue and the neonatal rat cardiomyocytes (NRCM). Its protein level was severely decreased after myocardial ischemia/reperfusion (I/R; 30 min/24 h) or hypoxia/reoxygenation (H/R; 24 h/4 h). Claudin-5 was present in the mitochondria of NRCM as determined by confocal microscopy. H/R-induced downregulation of claudin-5 was accompanied by mitochondrial fragmentation. The protein level of mitofusin 2 (Mfn2) was dramatically decreased while the expression of dynamin-related protein (Drp) 1 was significantly increased after H/R. H/R-induced mitochondrial swelling and fission were observed by transmission electron microscope (TEM). Overexpression of claudin-5 by adenoviral infection reversed these structural disintegration of mitochondria. The mitochondria-centered intrinsic pathway of apoptosis triggered by H/R and indicated by the expression of cytochrome c and cleaved caspase 3 in the cytoplasm of NRCMs was also reduced by overexpressing claudin-5. Overexpression of claudin-5 in mouse heart also significantly decreased cleaved caspase 3 expression and the infarct size in ischemic heart with improved systolic function. CONCLUSION We demonstrated for the first time the presence of claudin-5 in the mitochondria in cardiomyocytes and provided the firm evidence for the cardioprotective role of claudin-5 in the preservation of mitochondrial dynamics and cell fate against hypoxia- or ischemia-induced stress.
Collapse
Affiliation(s)
- Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Division of Cardiology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA.
| | - Haiqiong Liu
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Baihe Chen
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Division of Cardiology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Han Liu
- Division of Cardiology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Ahmed Abdel-Latif
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, 40536-0509, USA
| | - Masafumi Kitakaze
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, 5675-8565, Japan
| | - Xianbao Wang
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Yuanzhou Wu
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Dylan Chou
- Department of Physiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Jin Kyung Kim
- Division of Cardiology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
15
|
Zou R, Tao J, Qiu J, Shi W, Zou M, Chen W, Li W, Zhou N, Wang S, Ma L, Chen X. Ndufs1 Deficiency Aggravates the Mitochondrial Membrane Potential Dysfunction in Pressure Overload-Induced Myocardial Hypertrophy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5545261. [PMID: 33763166 PMCID: PMC7952157 DOI: 10.1155/2021/5545261] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/03/2021] [Accepted: 02/18/2021] [Indexed: 12/11/2022]
Abstract
Mitochondrial dysfunction has been suggested to be the key factor in the development and progression of cardiac hypertrophy. The onset of mitochondrial dysfunction and the mechanisms underlying the development of cardiac hypertrophy (CH) are incompletely understood. The present study is based on the use of multiple bioinformatics analyses for the organization and analysis of scRNA-seq and microarray datasets from a transverse aortic constriction (TAC) model to examine the potential role of mitochondrial dysfunction in the pathophysiology of CH. The results showed that NADH:ubiquinone oxidoreductase core subunit S1- (Ndufs1-) dependent mitochondrial dysfunction plays a key role in pressure overload-induced CH. Furthermore, in vivo animal studies using a TAC mouse model of CH showed that Ndufs1 expression was significantly downregulated in hypertrophic heart tissue compared to that in normal controls. In an in vitro model of angiotensin II- (Ang II-) induced cardiomyocyte hypertrophy, Ang II treatment significantly downregulated the expression of Ndufs1 in cardiomyocytes. In vitro mechanistic studies showed that Ndufs1 knockdown induced CH; decreased the mitochondrial DNA content, mitochondrial membrane potential (MMP), and mitochondrial mass; and increased the production of mitochondrial reactive oxygen species (ROS) in cardiomyocytes. On the other hand, Ang II treatment upregulated the expression levels of atrial natriuretic peptide, brain natriuretic peptide, and myosin heavy chain beta; decreased the mitochondrial DNA content, MMP, and mitochondrial mass; and increased mitochondrial ROS production in cardiomyocytes. The Ang II-mediated effects were significantly attenuated by overexpression of Ndufs1 in rat cardiomyocytes. In conclusion, our results demonstrate downregulation of Ndufs1 in hypertrophic heart tissue, and the results of mechanistic studies suggest that Ndufs1 deficiency may cause mitochondrial dysfunction in cardiomyocytes, which may be associated with the development and progression of CH.
Collapse
Affiliation(s)
- Rongjun Zou
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Wanting Shi
- Department of Paediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Minghui Zou
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Weidan Chen
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Wenlei Li
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Na Zhou
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Shaoli Wang
- Department of Surgical Nursing, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Li Ma
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xinxin Chen
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| |
Collapse
|
16
|
Lou T, Ma J, Xie Y, Yao G, Fan Y, Ma S, Zou X. Nuanxin capsule enhances cardiac function by inhibiting oxidative stress-induced mitochondrial dependent apoptosis through AMPK/JNK signaling pathway. Biomed Pharmacother 2021; 135:111188. [PMID: 33418304 DOI: 10.1016/j.biopha.2020.111188] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Oxidative stress and apoptosis play critical roles in the pathogenesis of heart failure (HF).Nuanxin capsule (NX) is a Chinese medicine that has outstanding protective effects on HF. The present study aimed to elucidate whether NX could protect HF against oxidative stress-induced apoptosis through intrinsic mitochondrial pathway. METHODS In vivo, HF was induced by transverse aortic constriction. NX and Compound C (Comp C) were administered to C57BL/6 J mice for over a 4-week period. Cardiac function was assessed with echocardiography. In vitro, H9c2 cells were exposed to H2O2 in the presence or absence of NX and Compound C. Cell viability, cytotoxicity, reactive oxygen species (ROS) production, apoptosis, mitochondrial membrane potential (ΔΨm) and mitochondrial function by oxygen consumption rate (OCR) were detected. The expressions of cytochrome c, BAX, Bcl-2, cleaved caspase-3, AMPK and JNK were evaluated by western blotting. RESULTS The results indicated that NX significantly improved cardiac function and enhanced the cell viability, ΔΨm and mitochondrial respiration. Also NX treatment reduced cell cytotoxicity and ROS production. Moreover, NX inhibited mitochondrial-mediated apoptosis by upregulating AMPK and downregulating JNK both in vivo and in vitro. The protective effects of NX on cardiac function by reducing oxidative stress-induced mitochondrial dependent apoptosis were reversed by Compound C treatment. CONCLUSIONS These findings demonstrated that NX effectively improved cardiac function in TAC mice by reducing oxidative stress-induced mitochondrial dependent apoptosis by activating AMPK/JNK signaling pathway.
Collapse
Affiliation(s)
- Tiantian Lou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Jin Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Yanzheng Xie
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Gengzhen Yao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Ye Fan
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China
| | - Shiyu Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China.
| | - Xu Zou
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, China; Dongguan Kanghua Hospital, Dongguan, 523080, China.
| |
Collapse
|
17
|
Malfatto G, Villani A, Parati G. Heart failure features and sacubitril/valsartan effects. J Cardiovasc Med (Hagerstown) 2021; 22:234-235. [PMID: 33136813 DOI: 10.2459/jcm.0000000000001128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Gabriella Malfatto
- Istituto Auxologico Italiano IRCCS, dipartimento di Scienze Cardiovascolari, Neurologiche , metaboliche, ospedale S.Luca, Milano, Italy
| | - Alessandra Villani
- Istituto Auxologico Italiano IRCCS, dipartimento di Scienze Cardiovascolari, Neurologiche , metaboliche, ospedale S.Luca, Milano, Italy
| | - Gianfranco Parati
- Dipartimento di Medicina e Chirurgia, Università di Milano-Bicocca, Milano, Italy
| |
Collapse
|
18
|
Gerber L, Clow KA, Mark FC, Gamperl AK. Improved mitochondrial function in salmon (Salmo salar) following high temperature acclimation suggests that there are cracks in the proverbial 'ceiling'. Sci Rep 2020; 10:21636. [PMID: 33303856 PMCID: PMC7729908 DOI: 10.1038/s41598-020-78519-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/22/2020] [Indexed: 11/09/2022] Open
Abstract
Mitochondrial function can provide key insights into how fish will respond to climate change, due to its important role in heart performance, energy metabolism and oxidative stress. However, whether warm acclimation can maintain or improve the energetic status of the fish heart when exposed to short-term heat stress is not well understood. We acclimated Atlantic salmon, a highly aerobic eurythermal species, to 12 and 20 °C, then measured cardiac mitochondrial functionality and integrity at 20 °C and at 24, 26 and 28 °C (this species' critical thermal maximum ± 2 °C). Acclimation to 20 °C vs. 12 °C enhanced many aspects of mitochondrial respiratory capacity and efficiency up to 24 °C, and preserved outer mitochondrial membrane integrity up to 26 °C. Further, reactive oxygen species (ROS) production was dramatically decreased at all temperatures. These data suggest that salmon acclimated to 'normal' maximum summer temperatures are capable of surviving all but the most extreme ocean heat waves, and that there is no 'tradeoff' in heart mitochondrial function when Atlantic salmon are acclimated to high temperatures (i.e., increased oxidative phosphorylation does not result in heightened ROS production). This study suggests that fish species may show quite different acclimatory responses when exposed to prolonged high temperatures, and thus, susceptibility to climate warming.
Collapse
Affiliation(s)
- Lucie Gerber
- Department of Ocean Sciences, Memorial University, St. John's, NL, Canada.
| | - Kathy A Clow
- Department of Ocean Sciences, Memorial University, St. John's, NL, Canada
| | - Felix C Mark
- Section Integrative Ecophysiology, Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, Bremerhaven, Germany
| | - Anthony K Gamperl
- Department of Ocean Sciences, Memorial University, St. John's, NL, Canada
| |
Collapse
|
19
|
Comparison of the Effect of Sacubitril/Valsartan on Left Ventricular Systolic Function in Patients with Non-ischaemic and Ischaemic Cardiomyopathy. Cardiovasc Drugs Ther 2020; 34:755-762. [DOI: 10.1007/s10557-020-07036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/05/2020] [Indexed: 10/23/2022]
|
20
|
Thompson J, Maceyka M, Chen Q. Targeting ER stress and calpain activation to reverse age-dependent mitochondrial damage in the heart. Mech Ageing Dev 2020; 192:111380. [PMID: 33045249 DOI: 10.1016/j.mad.2020.111380] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/17/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Severity of cardiovascular disease increases markedly in elderly patients. In addition, many therapeutic strategies that decrease cardiac injury in adult patients are invalid in elderly patients. Thus, it is a challenge to protect the aged heart in the context of underlying chronic or acute cardiac diseases including ischemia-reperfusion injury. The cause(s) of this age-related increased damage remain unknown. Aging impairs the function of the mitochondrial electron transport chain (ETC), leading to decreased energy production and increased oxidative stress due to generation of reactive oxygen species (ROS). Additionally, ROS-induced oxidative stress can increase cardiac injury during ischemia-reperfusion by potentiating mitochondrial permeability transition pore (MPTP) opening. Aging leads to increased endoplasmic reticulum (ER) stress, which contributes to mitochondrial dysfunction, including reduced function of the ETC. The activation of both cytosolic and mitochondrial calcium-activated proteases termed calpains leads to mitochondrial dysfunction and decreased ETC function. Intriguingly, mitochondrial ROS generation also induces ER stress, highlighting the dynamic interaction between mitochondria and ER. Here, we discuss the role of ER stress in sensitizing and potentiating mitochondrial dysfunction in response to ischemia-reperfusion, and the promising potential therapeutic benefit of inhibition of ER stress and / or calpains to attenuate cardiac injury in elderly patients.
Collapse
Affiliation(s)
- Jeremy Thompson
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, United States
| | - Michael Maceyka
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, United States
| | - Qun Chen
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, United States.
| |
Collapse
|
21
|
Yarmohammadi F, Rezaee R, Karimi G. Natural compounds against doxorubicin-induced cardiotoxicity: A review on the involvement of Nrf2/ARE signaling pathway. Phytother Res 2020; 35:1163-1175. [PMID: 32985744 DOI: 10.1002/ptr.6882] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/01/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
Cardiotoxicity is the main concern for long-term use of the doxorubicin (DOX). Reactive oxygen species (ROS) generation leads to oxidative stress that significantly contributes to the cardiac damage induced by DOX. The nuclear factor erythroid 2-related factor (Nrf2) acts as a protective player against DOX-induced myocardial oxidative stress. Several natural compounds (NCs) with anti-oxidative effects, were examined to suppress DOX cardiotoxicity such as asiatic acid, α-linolenic acid, apigenin, baicalein, β-lapachone, curdione, dioscin, ferulic acid, Ganoderma lucidum polysaccharides, genistein, ginsenoside Rg3, indole-3-carbinol, naringenin-7-O-glucoside, neferine, p-coumaric acid, pristimerin, punicalagin, quercetin, sulforaphane, and tanshinone IIA. The present article, reviews NCs that showed protective effects against DOX-induced cardiac injury through induction of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
22
|
Ioannou A, Metaxa S, Simon S, Mandal AKJ, Missouris CG. Letter to the Editor - New Pharmacotherapy for Heart Failure with Reduced Ejection Fraction. Expert Rev Cardiovasc Ther 2020; 18:651-652. [PMID: 32744124 DOI: 10.1080/14779072.2020.1799562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Adam Ioannou
- Department of Cardiology, Royal Free Hospital, Royal Free NHS Foundation Trust , London, UK
| | - Sofia Metaxa
- Department of Cardiology, Wrexham Park Hospital, Frimley Health NHS Foundation Trust , Slough, UK
| | - Steny Simon
- Department of Cardiology, Wrexham Park Hospital, Frimley Health NHS Foundation Trust , Slough, UK
| | - Amit K J Mandal
- Department of Cardiology, Wrexham Park Hospital, Frimley Health NHS Foundation Trust , Slough, UK
| | - Constantinos G Missouris
- Department of Cardiology, Wrexham Park Hospital, Frimley Health NHS Foundation Trust , Slough, UK.,Medical School, University of Cyprus Medical School , Nicosia, Cyprus
| |
Collapse
|
23
|
Ruiz-Meana M, Bou-Teen D, Ferdinandy P, Gyongyosi M, Pesce M, Perrino C, Schulz R, Sluijter JPG, Tocchetti CG, Thum T, Madonna R. Cardiomyocyte ageing and cardioprotection: consensus document from the ESC working groups cell biology of the heart and myocardial function. Cardiovasc Res 2020; 116:1835-1849. [DOI: 10.1093/cvr/cvaa132] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/25/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Advanced age is a major predisposing risk factor for the incidence of coronary syndromes and comorbid conditions which impact the heart response to cardioprotective interventions. Advanced age also significantly increases the risk of developing post-ischaemic adverse remodelling and heart failure after ischaemia/reperfusion (IR) injury. Some of the signalling pathways become defective or attenuated during ageing, whereas others with well-known detrimental consequences, such as glycoxidation or proinflammatory pathways, are exacerbated. The causative mechanisms responsible for all these changes are yet to be elucidated and are a matter of active research. Here, we review the current knowledge about the pathophysiology of cardiac ageing that eventually impacts on the increased susceptibility of cells to IR injury and can affect the efficiency of cardioprotective strategies.
Collapse
Affiliation(s)
- Marisol Ruiz-Meana
- Department of Cardiology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autonoma de Barcelona and Centro de Investigación Biomédica en Red-CV, CIBER-CV, Madrid, Spain
| | - Diana Bou-Teen
- Department of Cardiology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autonoma de Barcelona and Centro de Investigación Biomédica en Red-CV, CIBER-CV, Madrid, Spain
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Mariann Gyongyosi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Joost P G Sluijter
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Department of Internal Medicine, University of Texas Medical School in Houston, Houston, TX, USA
| |
Collapse
|
24
|
Báez AL, Lo Presti MS, Bazán PC, Strauss M, Velázquez López DA, Miler N, Rivarola HW, Paglini-Oliva PA. Analysis of mitochondrial enzymatic activity in blood lymphomonocyte fractions during infection with different Trypanosoma cruzi strains. Rev Inst Med Trop Sao Paulo 2020; 62:e15. [PMID: 32074218 PMCID: PMC7032009 DOI: 10.1590/s1678-9946202062015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/02/2019] [Indexed: 01/12/2023] Open
Abstract
Proinflammatory and inflammatory mediators induced by Trypanosoma
cruzi infection increase the oxidative stress, generating toxicity
for cells targeting mitochondria of different tissues. We studied the activity
of citrate synthase and complexes I-IV of respiratory chain in mitochondria of
blood lymphomonocyte fraction, from albino Swiss mice infected with different
isolates of T. cruzi, during Chagas disease evolution.
Complexes I-IV were modified in infected groups (p<0.05) in all the stages,
and an inflammatory process of different magnitudes was detected in the heart
and skeletal muscle according to the isolate. The citrate synthase activity
presented modifications in the SGO Z12 and the Tulahuen group (p<0.05).
Hearts showed fiber fragmentation and fibrosis; skeletal muscle presented
inflammatory infiltrates and in the Tulahuen infected group, there were also
amastigote nests. The inflammatory processes produced an oxidative stress that
induced different alterations of mitochondrial enzymes activities in the
lymphomonocyte fraction that can be detected by a simple blood extraction,
suggesting that they could be used as disease markers, especially in the
indeterminate phase of Chagas disease.
Collapse
Affiliation(s)
- Alejandra L Báez
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - María S Lo Presti
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - Paola C Bazán
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - Mariana Strauss
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - Daniela A Velázquez López
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - Noemí Miler
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - Héctor W Rivarola
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| | - Patricia A Paglini-Oliva
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, INICSA-CONICET, Córdoba, Argentina
| |
Collapse
|
25
|
No MH, Heo JW, Yoo SZ, Kim CJ, Park DH, Kang JH, Seo DY, Han J, Kwak HB. Effects of aging and exercise training on mitochondrial function and apoptosis in the rat heart. Pflugers Arch 2020; 472:179-193. [PMID: 32048000 DOI: 10.1007/s00424-020-02357-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/18/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022]
Abstract
Aging is associated with vulnerability to cardiovascular diseases, and mitochondrial dysfunction plays a critical role in cardiovascular disease pathogenesis. Exercise training is associated with benefits against chronic cardiac diseases. The purpose of this study was to determine the effects of aging and treadmill exercise training on mitochondrial function and apoptosis in the rat heart. Fischer 344 rats were divided into young sedentary (YS; n = 10, 4 months), young exercise (YE; n = 10, 4 months), old sedentary (OS; n = 10, 20 months), and old exercise (OE; n = 10, 20 months) groups. Exercise training groups ran on a treadmill at 15 m/min (young) or 10 m/min (old), 45 min/day, 5 days/week for 8 weeks. Morphological parameters, mitochondrial function, mitochondrial dynamics, mitophagy, and mitochondria-mediated apoptosis were analyzed in cardiac muscle. Mitochondrial O2 respiratory capacity and Ca2+ retention capacity gradually decreased, and mitochondrial H2O2 emitting potential significantly increased with aging. Exercise training attenuated aging-induced mitochondrial H2O2 emitting potential and mitochondrial O2 respiratory capacity, while protecting Ca2+ retention in the old groups. Aging triggered imbalanced mitochondrial dynamics and excess mitophagy, while exercise training ameliorated the aging-induced imbalance in mitochondrial dynamics and excess mitophagy. Aging induced increase in Bax and cleaved caspase-3 protein levels, while decreasing Bcl-2 levels. Exercise training protected against the elevation of apoptotic signaling markers by decreasing Bax and cleaved caspase-3 and increasing Bcl-2 protein levels, while decreasing the Bax/Bcl-2 ratio and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive myonuclei. These data demonstrate that regular exercise training prevents aging-induced impairment of mitochondrial function and mitochondria-mediated apoptosis in cardiac muscles.
Collapse
Affiliation(s)
- Mi-Hyun No
- Department of Kinesiology, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Jun-Won Heo
- Department of Kinesiology, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Su-Zi Yoo
- Department of Kinesiology, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, Kyung Hee University, Seoul, South Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology and Medicinal Toxicology Research Center, Inha University School of Medicine, Incheon, South Korea
| | - Dae-Yun Seo
- Department of Physiology and Cardiovascular and Metabolic Disease Center, Inje University School of Medicine, Busan, South Korea
| | - Jin Han
- Department of Physiology and Cardiovascular and Metabolic Disease Center, Inje University School of Medicine, Busan, South Korea
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea.
| |
Collapse
|
26
|
Gen WR, Fu CY, He HH, Zheng MZ, Wang LL, Yang Y, Shen YL, Chen YY. Linagliptin improved myocardial function recovery in rat hearts after a prolonged hypothermic preservation. Life Sci 2018; 210:47-54. [PMID: 30170072 DOI: 10.1016/j.lfs.2018.08.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/17/2018] [Accepted: 08/26/2018] [Indexed: 11/18/2022]
Abstract
AIMS To determine whether linagliptin, a dipeptidyl peptidase 4 inhibitor, can promote the recovery of cardiac function after hypothermic preservation. MAIN METHODS Rat hearts were preserved in cold Celsior solution with or without linagliptin for 9 h. Cardiac function was evaluated at 60 min of reperfusion after hypothermic preservation. Cardiac mitochondrial morphology was observed using transmission electron microscope. The expression of dynamin-related protein 1 (Drp1), NADPH oxidase 2 (NOX2), calmodulin-dependent protein kinase II (CaMKII) were detected using Western blot. KEY FINDINGS Compared with Celsior group, supplement of Celsior solution with linagliptin (0.25-0.75 nM) could significantly prevent hypothermic preservation-induced cardiac dysfunction. The expression of NOX2 protein, ROS level and MDA content in cardium were increased after hypothermic preservation, which was inhibited by linagliptin. Although the mitofusin1, 2, optic atrophy type 1, and total Drp1 expression in myocardium did not change, the level of p-Drp1 S616 and mitochondrial Drp1 were enhanced after hypothermic preservation. Linagliptin supplement could inhibit the hypothermic preservation-induced increase in p-Drp1 S616 and mitochondrial Drp1 protein, and mitigate the mitochondrial fragmentation. Level of p-CaMKII protein enhanced after hypothermic preservation, which could be prevented by linagliptin or a NOX2 inhibitor Phox-I2. Both Phox-I2 and a CaMKII inhibitor KN-93 could reduce the hypothermic preservation-induced increase in p-Drp1 S616 and mitochondrial Drp1 protein. SIGNIFICANCE Supplement Celsior solution with linagliptin could improve cardiac function recovery in 9-h hypothermic preserved rat hearts. The cardioprotective effect of linagliptin might be due to the inhibition of Drp1 phosphorylation and mitochondrial translocation by preventing NOX2-mediated CaMKII activation.
Collapse
Affiliation(s)
- Wei-Ran Gen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chun-Yan Fu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hui-Hui He
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ming-Zhi Zheng
- Department of Pharmacology, Hangzhou Medical College, Hangzhou 310053, China
| | - Lin-Lin Wang
- Center for Stem Cell and Tissue Engineering, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yi Yang
- Department of Pharmacology, Medical School of Jinhua Polytechnic, Jinhua 321007, China
| | - Yue-Liang Shen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Ying-Ying Chen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
27
|
Lu Z, Cui Y, Wei X, Gao P, Zhang H, Wei X, Li Q, Sun F, Yan Z, Zheng H, Yang G, Liu D, Zhu Z. Deficiency of PKD2L1 (TRPP3) Exacerbates Pathological Cardiac Hypertrophy by Augmenting NCX1-Mediated Mitochondrial Calcium Overload. Cell Rep 2018; 24:1639-1652. [DOI: 10.1016/j.celrep.2018.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/19/2018] [Accepted: 07/06/2018] [Indexed: 10/28/2022] Open
|
28
|
Dey S, DeMazumder D, Sidor A, Foster DB, O'Rourke B. Mitochondrial ROS Drive Sudden Cardiac Death and Chronic Proteome Remodeling in Heart Failure. Circ Res 2018; 123:356-371. [PMID: 29898892 DOI: 10.1161/circresaha.118.312708] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE Despite increasing prevalence and incidence of heart failure (HF), therapeutic options remain limited. In early stages of HF, sudden cardiac death (SCD) from ventricular arrhythmias claims many lives. Reactive oxygen species (ROS) have been implicated in both arrhythmias and contractile dysfunction. However, little is known about how ROS in specific subcellular compartments contribute to HF or SCD pathophysiology. The role of ROS in chronic proteome remodeling has not been explored. OBJECTIVE We will test the hypothesis that elevated mitochondrial ROS (mROS) is a principal source of oxidative stress in HF and in vivo reduction of mROS mitigates SCD. METHODS AND RESULTS Using a unique guinea pig model of nonischemic HF that recapitulates important features of human HF, including prolonged QT interval and high incidence of spontaneous arrhythmic SCD, compartment-specific ROS sensors revealed increased mROS in resting and contracting left ventricular myocytes in failing hearts. Importantly, the mitochondrially targeted antioxidant (MitoTEMPO) normalized global cellular ROS. Further, in vivo MitoTEMPO treatment of HF animals prevented and reversed HF, eliminated SCD by decreasing dispersion of repolarization and ventricular arrhythmias, suppressed chronic HF-induced remodeling of the expression proteome, and prevented specific phosphoproteome alterations. Pathway analysis of mROS-sensitive networks indicated that increased mROS in HF disrupts the normal coupling between cytosolic signals and nuclear gene programs driving mitochondrial function, antioxidant enzymes, Ca2+ handling, and action potential repolarization, suggesting new targets for therapeutic intervention. CONCLUSIONS mROS drive both acute emergent events, such as electrical instability responsible for SCD, and those that mediate chronic HF remodeling, characterized by suppression or altered phosphorylation of metabolic, antioxidant, and ion transport protein networks. In vivo reduction of mROS prevents and reverses electrical instability, SCD, and HF. Our findings support the feasibility of targeting the mitochondria as a potential new therapy for HF and SCD while identifying new mROS-sensitive protein modifications.
Collapse
Affiliation(s)
- Swati Dey
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (S.D., D.D., A.S., D.B.F., B.O.)
| | - Deeptankar DeMazumder
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (S.D., D.D., A.S., D.B.F., B.O.).,Division of Cardiology, Department of Medicine, University of Cincinnati, OH (D.D.)
| | - Agnieszka Sidor
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (S.D., D.D., A.S., D.B.F., B.O.)
| | - D Brian Foster
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (S.D., D.D., A.S., D.B.F., B.O.)
| | - Brian O'Rourke
- From the Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD (S.D., D.D., A.S., D.B.F., B.O.)
| |
Collapse
|
29
|
Lu YT, Li LZ, Yang YL, Yin X, Liu Q, Zhang L, Liu K, Liu B, Li J, Qi LW. Succinate induces aberrant mitochondrial fission in cardiomyocytes through GPR91 signaling. Cell Death Dis 2018; 9:672. [PMID: 29867110 PMCID: PMC5986788 DOI: 10.1038/s41419-018-0708-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/18/2018] [Accepted: 05/10/2018] [Indexed: 01/12/2023]
Abstract
Altered mitochondrial metabolism acts as an initial cause for cardiovascular diseases and metabolic intermediate succinate emerges as a mediator of mitochondrial dysfunction. This work aims to investigate whether or not extracellular succinate accumulation and its targeted G protein-coupled receptor-91 (GPR91) activation induce cardiac injury through mitochondrial impairment. The results showed that extracellular succinate promoted the translocation of dynamin-related protein 1 (Drp1) to mitochondria via protein kinase Cδ (PKCδ) activation, and induced mitochondrial fission factor (MFF) phosphorylation via extracellular signal-regulated kinases-1/2 (ERK1/2) activation in a GPR91-dependent manner. As a result, enhanced localization of MFF and Drp1 in mitochondria promoted mitochondrial fission, leading to mitochondrial dysfunction and cardiomyocyte apoptosis. We further showed that inhibition of succinate release and GPR91 signaling ameliorated oxygen-glucose deprivation-induced injury in cardiomyocytes and isoproterenol-induced myocardial ischemia injury in mice. Taken together, these results showed that in response to cardiac ischemia, succinate release activated GPR91 and induced mitochondrial fission via regulation of PKCδ and ERK1/2 signaling branches. These findings suggest that inhibition of extracellular succinate-mediated GPR91 activation might be a potential therapeutic strategy for protecting cardiomyocytes from ischemic injury.
Collapse
Affiliation(s)
- Yi-Tong Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lan-Zhu Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yi-Lin Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaojian Yin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qun Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Lei Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Kang Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Baolin Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Jia Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Lian-Wen Qi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
30
|
Lkhagva B, Kao YH, Lee TI, Lee TW, Cheng WL, Chen YJ. Activation of Class I histone deacetylases contributes to mitochondrial dysfunction in cardiomyocytes with altered complex activities. Epigenetics 2018; 13:376-385. [PMID: 29613828 DOI: 10.1080/15592294.2018.1460032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Histone deacetylases (HDACs) play vital roles in the pathophysiology of heart failure, which is associated with mitochondrial dysfunction. Tumor necrosis factor-α (TNF-α) contributes to the genesis of heart failure and impairs mitochondria. This study evaluated the role of HDACs in TNF-α-induced mitochondrial dysfunction and investigated their therapeutic potential and underlying mechanisms. We measured mitochondrial oxygen consumption rate (OCR) and ATP production using Seahorse XF24 extracellular flux analyzer and bioluminescent assay in control and TNF-α (10 ng/ml, 24 h)-treated HL-1 cells with or without HDAC inhibition. TNF-α increased Class I and II (but not Class IIa) HDAC activities (assessed by Luminescent) with enhanced expressions of Class I (HDAC1, HDAC2, HDAC3, and HDAC8) but not Class IIb HDAC (HDAC6 and HDAC10) proteins in HL-1 cells. TNF-α induced mitochondrial dysfunction with impaired basal, ATP-linked, and maximal respiration, decreased cellular ATP synthesis, and increased mitochondrial superoxide production (measured by MitoSOX red fluorescence), which were rescued by inhibiting HDACs with MPT0E014 (1 μM, a Class I and IIb inhibitor), or MS-275 (1 μM, a Class I inhibitor). MPT0E014 reduced TNF-α-decreased complex I and II enzyme (but not III or IV) activities (by enzyme activity microplate assays). Our results suggest that Class I HDAC actions contribute to TNF-α-induced mitochondrial dysfunction in cardiomyocytes with altered complex I and II enzyme regulation. HDAC inhibition improves dysfunctional mitochondrial bioenergetics with attenuation of TNF-α-induced oxidative stress, suggesting the therapeutic potential of HDAC inhibition in cardiac dysfunction.
Collapse
Affiliation(s)
- Baigalmaa Lkhagva
- a Graduate Institute of Clinical Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan.,b Department of Cardiology, School of Medicine , Mongolian National University of Medical Sciences , Ulaanbaatar , Mongolia
| | - Yu-Hsun Kao
- a Graduate Institute of Clinical Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan.,c Department of Medical Education and Research , Wan Fang Hospital , Taipei Medical University , Taipei , Taiwan
| | - Ting-I Lee
- d Division of Endocrinology and Metabolism , Department of General Medicine , School of Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan.,e Department of Internal Medicine , Wan Fang Hospital , Taipei Medical University , Taipei , Taiwan
| | - Ting-Wei Lee
- a Graduate Institute of Clinical Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan.,f Division of Endocrinology and Metabolism, Department of Internal Medicine , Wan Fang Hospital , Taipei Medical University , Taipei , Taiwan
| | - Wan-Li Cheng
- a Graduate Institute of Clinical Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan
| | - Yi-Jen Chen
- a Graduate Institute of Clinical Medicine, College of Medicine , Taipei Medical University , Taipei , Taiwan.,g Division of Cardiovascular Medicine, Department of Internal Medicine , Wan Fang Hospital , Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
31
|
Christen F, Desrosiers V, Dupont-Cyr BA, Vandenberg GW, Le François NR, Tardif JC, Dufresne F, Lamarre SG, Blier PU. Thermal tolerance and thermal sensitivity of heart mitochondria: Mitochondrial integrity and ROS production. Free Radic Biol Med 2018; 116:11-18. [PMID: 29294390 DOI: 10.1016/j.freeradbiomed.2017.12.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/17/2017] [Accepted: 12/29/2017] [Indexed: 12/21/2022]
Abstract
Cardiac mitochondrial metabolism provides 90% of the ATP necessary for the contractile exertion of the heart muscle. Mitochondria are therefore assumed to play a pivotal role in heart failure (HF), cardiovascular disease and ageing. Heat stress increases energy metabolism and oxygen demand in tissues throughout the body and imposes a major challenge on the heart, which is suspected of being the first organ to fail during heat stress. The underlying mechanisms inducing heart failure are still unclear. To pinpoint the processes implicated in HF during heat stress, we measured mitochondrial respiration rates and hydrogen peroxide production of isolated Arctic char (Salvelinus alpinus) heart mitochondria at 4 temperatures: 10°C (acclimation), 15°C, 20°C and 25°C (just over critical maximum). We found that at temperature ranges causing the loss of an organism's general homeostasis (between 20°C and 25°C) and with a substrate combination close to physiological conditions, the heat-induced increase in mitochondrial oxygen consumption levels off. More importantly, at the same state, hydrogen peroxide efflux increased by almost 50%. In addition, we found that individuals with low mitochondrial respiration rates produced more hydrogen peroxide at 10°C, 15°C and 20°C. This could indicate that individuals with cardiac mitochondria having a low respiratory capacity, have a more fragile heart and will be more prone to oxidative stress and HF, and less tolerant to temperature changes and other stressors. Our results show that, at temperatures close to the thermal limit, mitochondrial capacity is compromised and ROS production rates increase. This could potentially alter the performance of the cardiac muscle and lead to heat-induced HF underlining the important role that mitochondria play in setting thermal tolerance limits.
Collapse
Affiliation(s)
- Felix Christen
- Université du Québec à Rimouski, Département de biologie, Rimouski, Québec, Canada G5L3A1
| | - Véronique Desrosiers
- Université du Québec à Rimouski, Département de biologie, Rimouski, Québec, Canada G5L3A1
| | - Bernard A Dupont-Cyr
- Université du Québec à Rimouski, Département de biologie, Rimouski, Québec, Canada G5L3A1
| | - Grant W Vandenberg
- Université Laval, Département de sciences animales, Québec, Canada G1V 0A6
| | | | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, Montréal, Québec, Canada H1T 1C8
| | - France Dufresne
- Université du Québec à Rimouski, Département de biologie, Rimouski, Québec, Canada G5L3A1
| | - Simon G Lamarre
- Université de Moncton, Département de biologie, Moncton, New-Brunswick, Canada E1A 3E9
| | - Pierre U Blier
- Université du Québec à Rimouski, Département de biologie, Rimouski, Québec, Canada G5L3A1.
| |
Collapse
|
32
|
Szabo A, Sumegi K, Fekete K, Hocsak E, Debreceni B, Setalo G, Kovacs K, Deres L, Kengyel A, Kovacs D, Mandl J, Nyitrai M, Febbraio MA, Gallyas F, Sumegi B. Activation of mitochondrial fusion provides a new treatment for mitochondria-related diseases. Biochem Pharmacol 2018; 150:86-96. [PMID: 29378182 DOI: 10.1016/j.bcp.2018.01.038] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/22/2018] [Indexed: 02/07/2023]
Abstract
Mitochondria fragmentation destabilizes mitochondrial membranes, promotes oxidative stress and facilitates cell death, thereby contributing to the development and the progression of several mitochondria-related diseases. Accordingly, compounds that reverse mitochondrial fragmentation could have therapeutic potential in treating such diseases. BGP-15, a hydroxylamine derivative, prevents insulin resistance in humans and protects against several oxidative stress-related diseases in animal models. Here we show that BGP-15 promotes mitochondrial fusion by activating optic atrophy 1 (OPA1), a GTPase dynamin protein that assist fusion of the inner mitochondrial membranes. Suppression of Mfn1, Mfn2 or OPA1 prevents BGP-15-induced mitochondrial fusion. BGP-15 activates Akt, S6K, mTOR, ERK1/2 and AS160, and reduces JNK phosphorylation which can contribute to its protective effects. Furthermore, BGP-15 protects lung structure, activates mitochondrial fusion, and stabilizes cristae membranes in vivo determined by electron microscopy in a model of pulmonary arterial hypertension. These data provide the first evidence that a drug promoting mitochondrial fusion in in vitro and in vivo systems can reduce or prevent the progression of mitochondria-related disorders.
Collapse
Affiliation(s)
- Aliz Szabo
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Katalin Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Eniko Hocsak
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Balazs Debreceni
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Gyorgy Setalo
- Department of Medical Biology, University of Pécs Medical School, Pécs, Hungary; Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Laszlo Deres
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary; 1st Department of Medicine, Division of Cardiology, University of Pecs Medical School, Pecs, Hungary
| | - Andras Kengyel
- Department of Biophysics, University of Pécs Medical School, Pécs, Hungary
| | - Dominika Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Jozsef Mandl
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Miklos Nyitrai
- Department of Biophysics, University of Pécs Medical School, Pécs, Hungary
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary; Szentagothai Research Centre, University of Pécs, Pécs, Hungary; Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary; Szentagothai Research Centre, University of Pécs, Pécs, Hungary; Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
33
|
Milder degenerative effects of Carfilzomib vs. Bortezomib in the Drosophila model: a link to clinical adverse events. Sci Rep 2017; 7:17802. [PMID: 29259189 PMCID: PMC5736585 DOI: 10.1038/s41598-017-17596-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/27/2017] [Indexed: 12/15/2022] Open
Abstract
Proteasome inhibitors, e.g. Bortezomib (BTZ) and Carfilzomib (CFZ), have demonstrated clinical efficacy against haematological cancers. Interestingly, several adverse effects are less common, compared to BTZ, in patients treated with CFZ. As the molecular details of these observations remain not well understood we assayed the pathophysiological effects of CFZ vs. BTZ in the Drosophila experimental model. Mass Spectrometry analyses showed that neither CFZ nor BTZ are hydrolysed in flies’ tissues, while at doses inducing similar inhibition of the rate limiting for protein breakdown chymotrypsin-like (CT-L) proteasomal activity, CFZ treatment resulted in less intense increase of oxidative stress or activation of antioxidant and proteostatic modules. Also, despite comparable cardiotoxicity likely due to disrupted mitochondrial function, CFZ did not affect developmental processes, showed minimal neuromuscular defects and reduced to a lesser extent flies’ healthspan. Studies in flies, human cancer cell lines and blood cells isolated from Multiple Myeloma patients treated with CFZ or BTZ revealed, that the increased BTZ toxicity likely relates to partial co-inhibition of the caspase-like (C-L) proteasomal activity Supportively, co-treating flies with CFZ and a C-L selective proteasome inhibitor exacerbated CFZ-mediated toxicity. Our findings provide a reasonable explanation for the differential adverse effects of CFZ and BTZ in the clinic.
Collapse
|
34
|
Duni A, Liakopoulos V, Rapsomanikis KP, Dounousi E. Chronic Kidney Disease and Disproportionally Increased Cardiovascular Damage: Does Oxidative Stress Explain the Burden? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9036450. [PMID: 29333213 PMCID: PMC5733207 DOI: 10.1155/2017/9036450] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/18/2017] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) patients are among the groups at the highest risk for cardiovascular disease and significantly shortened remaining lifespan. CKD enhances oxidative stress in the organism with ensuing cardiovascular damage. Oxidative stress in uremia is the consequence of higher reactive oxygen species (ROS) production, whereas attenuated clearance of pro-oxidant substances and impaired antioxidant defenses play a complementary role. The pathophysiological mechanism underlying the increased ROS production in CKD is at least partly mediated by upregulation of the intrarenal angiotensin system. Enhanced oxidative stress in the setting of the uremic milieu promotes enzymatic modification of circulating lipids and lipoproteins, protein carbamylation, endothelial dysfunction via disruption of nitric oxide (NO) pathways, and activation of inflammation, thus accelerating atherosclerosis. Left ventricular hypertrophy (LVH) and heart failure are hallmarks of CKD. NADPH oxidase activation, xanthine oxidase, mitochondrial dysfunction, and NO-ROS are the main oxidative pathways leading to LVH and the cardiorenal syndrome. Finally, a subset of antioxidant enzymes, the paraoxonases (PON), deserves special attention due to abundant clinical evidence accumulated regarding reduced serum PON1 activity in CKD as a contributor to the increased burden of cardiovascular disease. Future, meticulously designed studies are needed to assess the effects of antioxidant therapy on patients with CKD.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, Medical School of the University of Ioannina, Ioannina, Greece
| | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Evangelia Dounousi
- Department of Nephrology, Medical School of the University of Ioannina, Ioannina, Greece
| |
Collapse
|
35
|
Vacek JC, Behera J, George AK, Kamat PK, Kalani A, Tyagi N. Tetrahydrocurcumin ameliorates homocysteine-mediated mitochondrial remodeling in brain endothelial cells. J Cell Physiol 2017; 233:3080-3092. [PMID: 28833102 DOI: 10.1002/jcp.26145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 08/08/2017] [Indexed: 12/18/2022]
Abstract
Homocysteine (Hcy) causes endothelial dysfunction by inducing oxidative stress in most neurodegenerative disorders. This dysfunction is highly correlated with mitochondrial dynamics such as fusion and fission. However, there are no strategies to prevent Hcy-induced mitochondrial remodeling. Tetrahydrocurcumin (THC) is an anti-inflammatory and anti-oxidant compound. We hypothesized that THC may ameliorates Hcy-induced mitochondria remodeling in mouse brain endothelial cells (bEnd3) cells. bEnd3 cells were exposed to Hcy treatment in the presence or absence of THC. Cell viability and autophagic cell death were measured with MTT and MDC staining assay. Reactive oxygen species (ROS) production was determined using DCFH-DA staining by confocal microscopy. Autophagy flux was assessed using a conventional GFP-microtubule-associated protein 1 light chain 3 (LC3) dot assay. Interaction of phagophore marker LC-3 with mitochondrial receptor NIX was observed by confocal imaging. Mitochondrial fusion and fission were evaluated by western blot and RT-PCR. Our results demonstrated that Hcy resulted in cell toxicity in a dose-dependent manner and supplementation of THC prevented the detrimental effects of Hcy on cell survival. Furthermore, Hcy also upregulated fission marker (DRP-1), fusion marker (Mfn2), and autophagy marker (LC-3). Finally, we observed that Hcy activated mitochondrial specific phagophore marker (LC-3) and co-localized with the mitochondrial receptor NIX, as viewed by confocal microscopy. Pretreatment of bEnd3 with THC (15 μM) ameliorated Hcy-induced oxidative damage, mitochondrial fission/fusion, and mitophagy. Our studies strongly suggest that THC has beneficial effects on mitochondrial remodeling and could be developed as a potential therapeutic agent against hyperhomocysteinemia (HHcy) induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jonathan C Vacek
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky
| | - Jyotirmaya Behera
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky
| | - Akash K George
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky
| | - Pradip K Kamat
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky
| | - Anuradha Kalani
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
36
|
Dludla PV, Joubert E, Muller CJF, Louw J, Johnson R. Hyperglycemia-induced oxidative stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic acid-2- O-β-D-glucoside. Nutr Metab (Lond) 2017; 14:45. [PMID: 28702068 PMCID: PMC5504778 DOI: 10.1186/s12986-017-0200-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022] Open
Abstract
Diabetic patients are at an increased risk of developing heart failure when compared to their non-diabetic counter parts. Accumulative evidence suggests chronic hyperglycemia to be central in the development of myocardial infarction in these patients. At present, there are limited therapies aimed at specifically protecting the diabetic heart at risk from hyperglycemia-induced injury. Oxidative stress, through over production of free radical species, has been hypothesized to alter mitochondrial function and abnormally augment the activity of the NADPH oxidase enzyme system resulting in accelerated myocardial injury within a diabetic state. This has led to a dramatic increase in the exploration of plant-derived materials known to possess antioxidative properties. Several edible plants contain various natural constituents, including polyphenols that may counteract oxidative-induced tissue damage through their modulatory effects of intracellular signaling pathways. Rooibos, an indigenous South African plant, well-known for its use as herbal tea, is increasingly studied for its metabolic benefits. Prospective studies linking diet rich in polyphenols from rooibos to reduced diabetes associated cardiovascular complications have not been extensively assessed. Aspalathin, a flavonoid, and phenylpyruvic acid-2-O-β-D-glucoside, a phenolic precursor, are some of the major compounds found in rooibos that can ameliorate hyperglycemia-induced cardiomyocyte damage in vitro. While the latter has demonstrated potential to protect against cell apoptosis, the proposed mechanism of action of aspalathin is linked to its capacity to enhance the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression, an intracellular antioxidant response element. Thus, here we review literature on the potential cardioprotective properties of flavonoids and a phenylpropenoic acid found in rooibos against diabetes-induced oxidative injury.
Collapse
Affiliation(s)
- Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Elizabeth Joubert
- Plant Bioactives Group, Post-Harvest and Wine Technology Division, Agricultural Research Council (ARC) Infruitec- Nietvoorbij, Stellenbosch, South Africa.,Department of Food Science, Stellenbosch University, Stellenbosch, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, P.O. Box 19070, Tygerberg, 7505 South Africa.,Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
37
|
Xia Y, Chen Z, Chen A, Fu M, Dong Z, Hu K, Yang X, Zou Y, Sun A, Qian J, Ge J. LCZ696 improves cardiac function via alleviating Drp1-mediated mitochondrial dysfunction in mice with doxorubicin-induced dilated cardiomyopathy. J Mol Cell Cardiol 2017. [DOI: 10.1016/j.yjmcc.2017.06.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
38
|
Dong Z, Zhao P, Xu M, Zhang C, Guo W, Chen H, Tian J, Wei H, Lu R, Cao T. Astragaloside IV alleviates heart failure via activating PPARα to switch glycolysis to fatty acid β-oxidation. Sci Rep 2017; 7:2691. [PMID: 28578382 PMCID: PMC5457407 DOI: 10.1038/s41598-017-02360-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/10/2017] [Indexed: 01/01/2023] Open
Abstract
In heart failure (HF), energy metabolism pathway in cardiac muscle changes from fatty acid β-oxidation to glycolysis. However, the exact mechanism is unknown. Sarcoendoplasmic reticulum Ca2+α ATPase (SERCA) expression is downregulated and mitochondrial function is reduced in HF, perhaps partly due to a substantially reduced energy supply for excitation–contraction coupling resulting from a lower fatty acid β-oxidation rate. We investigated whether Astragaloside IV can activate peroxisome proliferator-activated receptor alpha (PPARα) to stimulate fatty acid β-oxidation and increase cardiac energy production, improving mitochondrial function and the efficiency of SERCA in HF. In pressure overload-induced HF mice and isolated hypertrophic myocardial cells, fatty acid β-oxidation and heart function were substantially strengthened following Astragaloside IV treatment, as demonstrated by the increased expression of PPARα and SERCA2a. In vitro, Astragaloside IV regulated energy metabolism by increasing ATP production and enhancing mitochondrial function, attributable to increased oxygen consumption and slightly increased mitochondrial Ca2+ uptake. In HF, Astragaloside IV switched glycolysis to fatty acid β-oxidation, as confirmed by reduced anaerobic glycolysis and an increased oxygen consumption ratio. These results suggest that Astragaloside IV can stimulate fatty acid β-oxidation and improve mitochondrial function, which may present a novel cardioprotective treatment that inhibits the progress of HF.
Collapse
Affiliation(s)
- Zhiwei Dong
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038, China
| | - Pei Zhao
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Ming Xu
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Baylor college of medicine, Houston, Texas, USA
| | - Wei Guo
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Huihua Chen
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Jing Tian
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Hongchang Wei
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Rong Lu
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Tongtong Cao
- Department of Pathology, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
39
|
Targeting Metabolic Modulation and Mitochondrial Dysfunction in the Treatment of Heart Failure. Diseases 2017; 5:diseases5020014. [PMID: 28933367 PMCID: PMC5547981 DOI: 10.3390/diseases5020014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 12/11/2022] Open
Abstract
Despite significant improvements in morbidity and mortality with current evidence-based pharmaceutical-based treatment of heart failure (HF) over the previous decades, the burden of HF remains high. An alternative approach is currently being developed, which targets myocardial energy efficiency and the dysfunction of the cardiac mitochondria. Emerging evidence suggests that the insufficient availability of ATP to the failing myocardium can be attributed to abnormalities in the myocardial utilisation of its substrates rather than an overall lack of substrate availability. Therefore, the development of potential metabolic therapeutics has commenced including trimetazidine, ranolazine and perhexiline, as well as specific mitochondrial-targeting pharmaceuticals, such as elamipretide. Large randomised controlled trials are required to confirm the role of metabolic-modulating drugs in the treatment of heart failure, but early studies have been promising in their possible efficacy for the management of heart failure in the future.
Collapse
|
40
|
Riba A, Deres L, Eros K, Szabo A, Magyar K, Sumegi B, Toth K, Halmosi R, Szabados E. Doxycycline protects against ROS-induced mitochondrial fragmentation and ISO-induced heart failure. PLoS One 2017; 12:e0175195. [PMID: 28384228 PMCID: PMC5383248 DOI: 10.1371/journal.pone.0175195] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/22/2017] [Indexed: 11/18/2022] Open
Abstract
In addition to their anti-bacterial action, tetracyclines also have complex biological effects, including the modification of mitochondrial protein synthesis, metabolism and gene-expression. Long-term clinical studies have been performed using tetracyclines, without significant side effects. Previous studies demonstrated that doxycycline (DOX), a major tetracyclin antibiotic, exerted a protective effect in animal models of heart failure; however, its exact molecular mechanism is still unknown. Here, we provide the first evidence that DOX reduces oxidative stress-induced mitochondrial fragmentation and depolarization in H9c2 cardiomyocytes and beneficially alters the expression of Mfn-2, OPA-1 and Drp-1 -the main regulators of mitochondrial fusion and fission-in our isoproterenol (ISO)-induced heart failure model, ultimately decreasing the severity of heart failure. In mitochondria, oxidative stress causes a shift toward fission which leads to mitochondrial fragmentation and cell death. Protecting mitochondria from oxidative stress, and the regulation of mitochondrial dynamics by drugs that shift the balance toward fusion, could be a novel therapeutic approach for heart failure. On the basis of our findings, we raise the possibility that DOX could be a novel therapeutic agent in the future treatment of heart failure.
Collapse
Affiliation(s)
- Adam Riba
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
| | - Laszlo Deres
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
| | - Krisztian Eros
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
| | - Aliz Szabo
- Department of Biochemistry and Medical Chemistry, University of Pecs, Pécs, Hungary
| | - Klara Magyar
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
| | - Balazs Sumegi
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs, Pécs, Hungary
- MTA-PTE Nuclear-Mitochondrial Interactions Research Group, Pécs, Hungary
| | - Kalman Toth
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs, Pécs, Hungary
- MTA-PTE Nuclear-Mitochondrial Interactions Research Group, Pécs, Hungary
| | - Robert Halmosi
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
| | - Eszter Szabados
- 1st Department of Medicine, University of Pecs, Pécs, Hungary
- Szentagothai ResearchCenter, University of Pecs, Pécs, Hungary
| |
Collapse
|
41
|
Lesnefsky EJ, Chen Q, Tandler B, Hoppel CL. Mitochondrial Dysfunction and Myocardial Ischemia-Reperfusion: Implications for Novel Therapies. Annu Rev Pharmacol Toxicol 2017; 57:535-565. [PMID: 27860548 PMCID: PMC11060135 DOI: 10.1146/annurev-pharmtox-010715-103335] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria have emerged as key participants in and regulators of myocardial injury during ischemia and reperfusion. This review examines the sites of damage to cardiac mitochondria during ischemia and focuses on the impact of these defects. The concept that mitochondrial damage during ischemia leads to cardiac injury during reperfusion is addressed. The mechanisms that translate ischemic mitochondrial injury into cellular damage, during both ischemia and early reperfusion, are examined. Next, we discuss strategies that modulate and counteract these mechanisms of mitochondrial-driven injury. The new concept that mitochondria are not merely stochastic sites of oxidative and calcium-mediated injury but that they activate cellular responses of mitochondrial remodeling and cellular reactions that modulate the balance between cell death and recovery is reviewed, and the therapeutic implications of this concept are discussed.
Collapse
Affiliation(s)
- Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia 23298; ,
- Medical Service, McGuire Veterans Affairs Medical Center, Richmond, Virginia 23249;
| | - Qun Chen
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia 23298; ,
| | - Bernard Tandler
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, Ohio 44106;
| | - Charles L Hoppel
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106;
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
- Center for Mitochondrial Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| |
Collapse
|
42
|
Abstract
A core feature of ischemic heart disease is injury to cardiomyocytes (CMC). Ischemic CMC manifest the molecular mechanisms to undergo the major forms of cell injury and death, namely, oncotic necrosis, necroptosis, apoptosis and unregulated autophagy. Important modulators of ischemic injury are reperfusion and conditioning. Mitochondria have a major role in mediating the injury to CMC through membrane protein complexes referred to as death channels. Apoptosis is mediated by activation of a channel regulated by the Bcl-2 protein family leading to mitochondrial outer membrane permeabilization (MOMP). Oncotic type injury is mediated by opening of the mitochondrial permeability transition pore (mPTP). Mitochondria also have a reperfusion salvage kinase pathway (RISK). With cyclosporine A serving as a prototype, ongoing research is aimed at developing pharmacological approaches to condition and preserve mitochondrial integrity in order to promote CMC survival during episodes of myocardial ischemia.
Collapse
|
43
|
Adjusting MtDNA Quantification in Whole Blood for Peripheral Blood Platelet and Leukocyte Counts. PLoS One 2016; 11:e0163770. [PMID: 27736919 PMCID: PMC5063275 DOI: 10.1371/journal.pone.0163770] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/14/2016] [Indexed: 12/28/2022] Open
Abstract
Alterations of mitochondrial DNA copy number (mtDNAcn) in the blood (mitochondrial to nuclear DNA ratio) appear associated with several systemic diseases, including primary mitochondrial disorders, carcinogenesis, and hematologic diseases. Measuring mtDNAcn in DNA extracted from whole blood (WB) instead of from peripheral blood mononuclear cells or buffy coat may yield different results due to mitochondrial DNA present in platelets. The aim of this work is to quantify the contribution of platelets to mtDNAcn in whole blood [mtDNAcn(WB)] and to propose a correction formula to estimate leukocytes' mtDNAcn [mtDNAcn(L)] from mtDNAcn(WB). Blood samples from 10 healthy adults were combined with platelet-enriched plasma and saline solution to produce artificial blood preparations. Aliquots of each sample were combined with five different platelet concentrations. In 46 of these blood preparations, mtDNAcn was measured by qPCR. MtDNAcn(WB) increased 1.07 (95%CI 0.86, 1.29; p<0.001) per 1000 platelets present in the preparation. We proved that leukocyte count should also be taken into account as mtDNAcn(WB) was inversely associated with leukocyte count; it increased 1.10 (95%CI 0.95, 1.25, p<0.001) per unit increase of the ratio between platelet and leukocyte counts. If hematological measurements are available, subtracting 1.10 the platelets/leukocyte ratio from mtDNAcn(WB) may serve as an estimation for mtDNAcn(L). Both platelet and leukocyte counts in the sample are important sources of variation if comparing mtDNAcn among groups of patients when mtDNAcn is measured in DNA extracted from whole blood. Not taking the platelet/leukocyte ratio into account in whole blood measurements, may lead to overestimation and misclassification if interpreted as leukocytes' mtDNAcn.
Collapse
|
44
|
Ku HJ, Kwon OS, Kang BS, Lee DS, Lee HS, Park JW. IDH2 knockdown sensitizes tumor cells to emodin cytotoxicity in vitro and in vivo. Free Radic Res 2016; 50:1089-1097. [PMID: 27087448 DOI: 10.1080/10715762.2016.1178739] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although reactive oxygen species (ROS) work as second messengers at sublethal concentrations, higher levels of ROS can kill cancer cells. Since cellular ROS levels are determined by a balance between ROS generation and removal, the combination of ROS generators, and the depletion of reducing substances greatly enhance ROS levels. Emodin (1,3,8-trihydroxy-6-methyl anthraquinone), a natural anthraquinone derivative from the root and rhizome of numerous plants, is a ROS generator that induces apoptosis in cancer cells. The major enzyme to generate mitochondrial NADPH is the mitochondrial isoenzyme of NADP+-dependent isocitrate dehydrogenase (IDH2). In this report, we demonstrate that IDH2 knockdown effectively enhances emodin-induced apoptosis of mouse melanoma B16F10 cells through the regulation of ROS generation. Our findings suggest that suppression of IDH2 activity results in perturbation of the cellular redox balance and, ultimately, exacerbate emodin-induced apoptotic cell death in B16F10 cells. Our results strongly support a therapeutic strategy in the management of cancer that alters the intracellular redox status by the combination of a ROS generator and the suppression of antioxidant enzyme activity.
Collapse
Affiliation(s)
- Hyeong Jun Ku
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , College of Natural Sciences, Kyungpook National University , Taegu , Republic of Korea
| | - Oh-Shin Kwon
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , College of Natural Sciences, Kyungpook National University , Taegu , Republic of Korea
| | - Boem Sik Kang
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , College of Natural Sciences, Kyungpook National University , Taegu , Republic of Korea
| | - Dong-Seok Lee
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , College of Natural Sciences, Kyungpook National University , Taegu , Republic of Korea
| | - Hyun-Shik Lee
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , College of Natural Sciences, Kyungpook National University , Taegu , Republic of Korea
| | - Jeen-Woo Park
- a School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group , College of Natural Sciences, Kyungpook National University , Taegu , Republic of Korea
| |
Collapse
|
45
|
WITHDRAWN: Osthole attenuated myocardial ischemia/reperfusion via a mitochondrial apoptosis. Curr Res Transl Med 2016. [DOI: 10.1016/j.retram.2016.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
46
|
The Role of Mitochondrial Functional Proteins in ROS Production in Ischemic Heart Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5470457. [PMID: 27119006 PMCID: PMC4826939 DOI: 10.1155/2016/5470457] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/27/2016] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
Ischemic heart diseases (IHD) have become the leading cause of death around the world, killing more than 7 million people annually. In IHD, the blockage of coronary vessels will cause irreversible cell injury and even death. As the “powerhouse” and “apoptosis center” in cardiomyocytes, mitochondria play critical roles in IHD. Ischemia insult can reduce myocardial ATP content, resulting in energy stress and overproduction of reactive oxygen species (ROS). Thus, mitochondrial abnormality has been identified as a hallmark of multiple cardiovascular disorders. To date, many studies have suggested that these mitochondrial proteins, such as electron transport chain (ETC) complexes, uncoupling proteins (UCPs), mitochondrial dynamic proteins, translocases of outer membrane (Tom) complex, and mitochondrial permeability transition pore (MPTP), can directly or indirectly influence mitochondria-originated ROS production, consequently determining the degree of mitochondrial dysfunction and myocardial impairment. Here, the focus of this review is to summarize the present understanding of the relationship between some mitochondrial functional proteins and ROS production in IHD.
Collapse
|
47
|
Taegtmeyer H, Young ME, Lopaschuk GD, Abel ED, Brunengraber H, Darley-Usmar V, Des Rosiers C, Gerszten R, Glatz JF, Griffin JL, Gropler RJ, Holzhuetter HG, Kizer JR, Lewandowski ED, Malloy CR, Neubauer S, Peterson LR, Portman MA, Recchia FA, Van Eyk JE, Wang TJ. Assessing Cardiac Metabolism: A Scientific Statement From the American Heart Association. Circ Res 2016; 118:1659-701. [PMID: 27012580 DOI: 10.1161/res.0000000000000097] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In a complex system of interrelated reactions, the heart converts chemical energy to mechanical energy. Energy transfer is achieved through coordinated activation of enzymes, ion channels, and contractile elements, as well as structural and membrane proteins. The heart's needs for energy are difficult to overestimate. At a time when the cardiovascular research community is discovering a plethora of new molecular methods to assess cardiac metabolism, the methods remain scattered in the literature. The present statement on "Assessing Cardiac Metabolism" seeks to provide a collective and curated resource on methods and models used to investigate established and emerging aspects of cardiac metabolism. Some of those methods are refinements of classic biochemical tools, whereas most others are recent additions from the powerful tools of molecular biology. The aim of this statement is to be useful to many and to do justice to a dynamic field of great complexity.
Collapse
|
48
|
Yang L, Luo C, Chen C, Wang X, Shi W, Liu J. All-trans retinoic acid protects against doxorubicin-induced cardiotoxicity by activating the ERK2 signalling pathway. Br J Pharmacol 2015; 173:357-71. [PMID: 26507774 DOI: 10.1111/bph.13377] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/12/2015] [Accepted: 10/21/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Doxorubicin is a powerful antineoplastic agent for treating a wide range of cancers. However, doxorubicin cardiotoxicity of the heart has largely limited its clinical use. All-trans retinoic acid (ATRA) plays an important role in many cardiac biological processes, but its protective effects on doxorubicin-induced cardiotoxicity remain unknown. Here, we studied the effect of ATRA on doxorubicin cardiotoxicity and the underlying mechanisms. EXPERIMENTAL APPROACHES Cellular viability assays, Western blotting and mitochondrial respiration analyses were employed to evaluate the cellular response to ATRA in H9c2 cells and primary cardiomyocytes. Quantitative PCR and gene knockdown were performed to investigate the underlying molecular mechanisms of ATRA's effects on doxorubicin cardiotoxicity. KEY RESULTS ATRA significantly inhibited doxorubicin-induced apoptosis in H9c2 cells and primary cardiomyocytes. ATRA was more effective against doxorubicin cardiotoxicity than resveratrol and dexrazoxane. ATRA also suppressed reactive oxygen species generation and restored expression levels of mRNA and proteins in the phase II detoxifying enzyme system: nuclear factor-E2-related factor 2, manganese superoxide dismutase, haem oxygenase-1, and mitochondrial function (mitochondrial membrane integrity, mitochondrial DNA copy numbers and mitochondrial respiration capacity, biogenesis and dynamics). Both a ERK1/2 inhibitor (U0126) and ERK2 siRNA, but not ERK1 siRNA, abolished the protective effect of ATRA against doxorubicin-induced toxicity in H9c2 cells. Remarkably, ATRA did not compromise the anticancer efficacy of doxorubicin in gastric carcinoma cells. CONCLUSIONS AND IMPLICATIONS ATRA protected cardiomyocytes against doxorubicin-induced toxicity, by activating the ERK2 pathway, without compromising its anticancer efficacy. Therefore, ATRA is a promising candidate as a cardioprotective agent against doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Liang Yang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Cheng Luo
- School of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Cong Chen
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xun Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Wen Shi
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
49
|
Apelin regulates FoxO3 translocation to mediate cardioprotective responses to myocardial injury and obesity. Sci Rep 2015; 5:16104. [PMID: 26542760 PMCID: PMC4635427 DOI: 10.1038/srep16104] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/25/2015] [Indexed: 12/22/2022] Open
Abstract
The increasing incidence of obesity accentuates the importance of identifying mechanisms and optimal therapeutic strategies for patients with heart failure (HF) in relation to obesity status. Here, we investigated the association between plasma level of apelin, an adipocyte-derived factor, and clinicopathological features of obese and non-obese patients with HF. We further explored potential regulatory mechanisms of cardiac cell fate responses in conditions combining myocardial injury and obesity. In a prospective, cross-sectional study involving patients with HF we show that obese patients (BMI ≥ 30 kg/m(2)) have higher left ventricular ejection fraction (LVEF) and greater levels of plasma apelin (p < 0.005) than non-obese patients (< 30 kg/m(2)), independently of ischemic etiology. In a mouse model combining ischemia-reperfusion (I/R) injury and high-fat diet (HFD)-induced obesity, we identify apelin as a novel regulator of FoxO3 trafficking in cardiomyocytes. Confocal microscopy analysis of cardiac cells revealed that apelin prevents nuclear translocation of FoxO3 in response to oxygen deprivation through a PI3K pathway. These findings uncover apelin as a novel regulator of FoxO3 nucleocytoplasmic trafficking in cardiac cells in response to stress and provide insight into its potential clinical relevance in obese patients with HF.
Collapse
|
50
|
Dong D, Li L, Gu P, Jin T, Wen M, Yuan C, Gao X, Liu C, Zhang Z. Profiling metabolic remodeling in PP2Acα deficiency and chronic pressure overload mouse hearts. FEBS Lett 2015; 589:3631-9. [DOI: 10.1016/j.febslet.2015.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/08/2015] [Accepted: 10/11/2015] [Indexed: 01/02/2023]
|